1
|
Solís KH, Romero-Ávila MT, Alcántara-Hernández R, García-Sáinz JA. The many facets of biased signaling: Mechanisms and possible therapeutic implications. Pharmacol Ther 2025; 272:108877. [PMID: 40383400 DOI: 10.1016/j.pharmthera.2025.108877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/08/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
Receptor-mediated cell activation frequently results in a plethora of effects, and interestingly, not all agonists that act on a given receptor activate all of those actions to the same extent. Biased agonism refers to this fact, i.e., the possibility to activate only a part of the receptor's signaling capabilities. It is worth mentioning that Biased Signaling is an integral concept that includes the system (organisms, isolated tissues, or cells), the individual receptor studied, and the ligands. It should be remembered that the system's genetic expression profile defines the type, abundance, and cellular localization of proteins that participate in signaling. This short review will be focused on G protein receptors, but biased signaling occurs in many other receptor types. Biased signaling can be related to the G proteins and β-arrestins available. Similarly, enzymes that catalyze receptor posttranslational modifications, such as phosphorylation, acylation, or ubiquitination, can play a role. G protein-coupled receptor signaling occurs at the plasma membrane, but it is well-established that endosomal signaling is a functional reality. Therefore, paying attention to cellular elements that participate in receptor endosomal traffic and destination (recycling to the plasma membrane/ degradation) is pertinent. There is still much to be known about these bias mechanisms, which are essential for basic knowledge of receptor drug action and for treating many pathological entities.
Collapse
Affiliation(s)
- K Helivier Solís
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico
| | - M Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico.
| | - Rocío Alcántara-Hernández
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico.
| | - J Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria. Ap. Postal 70-600, Ciudad de México 04510. Mexico.
| |
Collapse
|
2
|
Guo R, Chen O, Zhou Y, Bang S, Chandra S, Li Y, Chen G, Xie RG, He W, Xu J, Zhou R, Song S, Person KL, Moore MN, Alwin AR, Spasojevic I, Jackson MR, Olson SH, Caron MG, Slosky LM, Wetsel WC, Barak LS, Ji RR. Arrestin-biased allosteric modulator of neurotensin receptor 1 alleviates acute and chronic pain. Cell 2025:S0092-8674(25)00508-2. [PMID: 40393456 DOI: 10.1016/j.cell.2025.04.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/12/2025] [Accepted: 04/28/2025] [Indexed: 05/22/2025]
Abstract
G-protein-biased agonists have been shown to enhance opioid analgesia by circumventing β-arrestin-2 (βarr2) signaling. We previously reported that SBI-553, a neurotensin receptor 1 (NTSR1)-positive allosteric modulator biased toward βarr2 signaling, attenuates psychostimulant effects in mice. Here, we demonstrate that its analog, SBI-810, exhibits potent antinociceptive properties in rodent models of postoperative pain, inflammatory pain, and neuropathic pain via systemic and local administration. SBI-810's analgesic effects require NTSR1 and βarr2 but not NTSR2 or βarr1. Mechanistically, SBI-810 suppresses excitatory synaptic transmission, inhibits NMDA receptor and extracellular-regulated signal kinase (ERK) signaling in spinal cord nociceptive neurons, reduces Nav1.7 surface expression and action potential firing in primary sensory neurons, and dampens C-fiber responses. Behaviorally, it reduces opioid-induced conditioned place preference, alleviates constipation, and mitigates chronic opioid withdrawal symptoms. These findings highlight NTSR1-biased allosteric modulators as a promising, non-addictive therapeutic strategy for acute and chronic pain management, acting through both peripheral and central mechanisms.
Collapse
Affiliation(s)
- Ran Guo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Ouyang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yang Zhou
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sangsu Bang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Sharat Chandra
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Yize Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Gang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Rou-Gang Xie
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Wei He
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Richard Zhou
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Shaoyong Song
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA
| | - Kelsey L Person
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Madelyn N Moore
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Abigail R Alwin
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Pharmacokinetics/Pharmacodynamics Core Laboratory, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael R Jackson
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Steven H Olson
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marc G Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lauren M Slosky
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - William C Wetsel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA; Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lawrence S Barak
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27705, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
3
|
Krumm BE, Roth BL. Intracellular GPCR modulators enable precision pharmacology. NPJ DRUG DISCOVERY 2025; 2:8. [PMID: 40371403 PMCID: PMC12069105 DOI: 10.1038/s44386-025-00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/20/2025] [Indexed: 05/16/2025]
Abstract
G-protein-coupled receptors (GPCRs) have proven to be the most successful target class for drug discovery but their complicated signal transduction pathways cause difficulties for drug development. Recently, ligands have been identified that engage an intracellular binding site which promotes pathway biased signal in cooperation with orthosteric ligands. Here, we explore the topic of biased signaling and intracellular modulators to understand their application for precision pharmacology of Class A or Rhodopsin-Like GPCRs.
Collapse
Affiliation(s)
- Brian E. Krumm
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Bryan L. Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- National Institute of Mental Health Psychoactive Drug Screening Program (NIMH PDSP), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| |
Collapse
|
4
|
Zhang J, Zhao X, Zhang L, Lu J, Bai J, An S, Zhu Y, Zhang H, Hao Y, Tian Y. Microbiota-derived acetate suppresses sympathetic outflow via olfactory receptor 59 in the rostral ventrolateral medulla. Pharmacol Res 2025; 216:107766. [PMID: 40345351 DOI: 10.1016/j.phrs.2025.107766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Dysbiosis of gut microbiota is closely associated with the development of hypertension, yet the underlying mechanism remains unclear. In this study, we aimed to elucidate the molecular mechanisms through which microbiota-derived acetate regulates sympathetic activity and arterial blood pressure (ABP). Bulk RNA sequencing was used to determine the expression of short-chain fatty acids receptors in the rostral ventrolateral medulla (RVLM). We examined the influence of olfactory receptor 59 (Olr59) on renal sympathetic nerve activity (RSNA) and ABP in anesthetized rats. The effect of Olr59 on the excitability of RVLM neurons was assessed through calcium imaging. Moreover, the 24-hour ambulatory blood pressure of conscious rats was recorded using radiotelemetry. The RNA sequencing results indicated that Olr59 was the most highly expressed short-chain fatty acids receptor in the rat RVLM and was upregulated in spontaneously hypertensive rats (SHR). Injection of the Olr59 agonist acetate or β-ionone into the RVLM reduced ABP and RSNA. However, administration of Olr59 antagonist or the knockdown of Olr59 in RVLM neurons did not significantly alter ABP, but it counteracted the hypotensive effect of acetate within the RVLM. Application of acetate or β-ionone to isolated brain slices mainly inhibited calcium signal in spinally-projecting RVLM neurons. Furthermore, overexpression of Olr59 in RVLM neurons via adeno-associated virus reduced ABP in SHR. Microbiota-derived acetate inhibits sympathetic activity and decreases blood pressure via Olr59 in the RVLM. Thus, Olr59 represents a promising new target for the treatment of hypertension.
Collapse
Affiliation(s)
- Jinye Zhang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xue Zhao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Lin Zhang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jinmeng Lu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jinlu Bai
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuo An
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yufang Zhu
- College of Nursing, Hebei Medical University, Shijiazhuang 050017, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang 050017, China
| | - Yinchao Hao
- Functional Laboratory, Experimental Center for Teaching, Hebei Medical University, Shijiazhuang 050017, China.
| | - Yanming Tian
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
5
|
Ma Y, Patterson B, Zhu L. Biased signaling in GPCRs: Structural insights and implications for drug development. Pharmacol Ther 2025; 266:108786. [PMID: 39719175 DOI: 10.1016/j.pharmthera.2024.108786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/18/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors in humans, playing a crucial role in regulating diverse cellular processes and serving as primary drug targets. Traditional drug design has primarily focused on ligands that uniformly activate or inhibit GPCRs. However, the concept of biased agonism-where ligands selectively stabilize distinct receptor conformations, leading to unique signaling outcomes-has introduced a paradigm shift in therapeutic development. Despite the promise of biased agonists to enhance drug efficacy and minimize side effects, a comprehensive understanding of the structural and biophysical mechanisms underlying biased signaling is essential. Recent advancements in GPCR structural biology have provided unprecedented insights into ligand binding, conformational dynamics, and the molecular basis of biased signaling. These insights, combined with improved techniques for characterizing ligand efficacy, have driven the development of biased ligands for several GPCRs, including opioid, angiotensin, and adrenergic receptors. This review synthesizes these developments, from mechanisms to drug discovery in biased signaling, emphasizing the role of structural insights in the rational design of next-generation biased agonists with superior therapeutic profiles. Ultimately, these advances hold the potential to revolutionize GPCR-targeted drug discovery, paving the way for more precise and effective treatments.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Brandon Patterson
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Lan Zhu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States.
| |
Collapse
|
6
|
Borah MP, Trakroo D, Soni N, Kumari P, Baidya M. Exploring Bias in GPCR Signaling and its Implication in Drug Development: A One-Sided Affair. Biochemistry 2025; 64:1-14. [PMID: 39613476 DOI: 10.1021/acs.biochem.4c00676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
G protein-coupled receptors (GPCRs) play a pivotal role in regulating numerous physiological processes through their interactions with two key effectors: G proteins and β-arrestins (βarrs). This makes them crucial targets for therapeutic drug development. Interestingly, the evolving concept of biased signaling where ligands selectively activate either the G proteins or the βarrs has not only refined our understanding of segregation of physiological responses downstream of GPCRs but has also revolutionized drug discovery, offering the potential for treatments with enhanced efficacy and minimal side effects. This Review explores the mechanisms behind biased agonism, exploring it through various lenses, including ligand, receptor, cellular systems, location, and tissue-specific biases. It also offers structural insights into both orthosteric and allosteric ligand-binding pockets, structural rearrangements associated with the loops, and how ligand-engineering can contribute to biased signaling. Moreover, we also discuss the unique conformational signature in an intrinsically biased GPCR, which currently remains relatively less explored and adds a new dimension in biased signaling. Lastly, we address the translational challenges and practical considerations in characterizing bias, emphasizing its therapeutic potential and the latest advancements in drug development. By designing ligands that target specific signaling pathways, biased signaling presents a transformative approach to creating safer and more effective therapies. This Review focuses on our current understanding of GPCR-biased signaling, discussing potential mechanisms that lead to bias, the effect of bias on GPCR structures at a molecular level, recent advancements, and its profound potential to drive innovation in drug discovery.
Collapse
Affiliation(s)
- Madhurjya Protim Borah
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jammu, Jammu, Jammu and Kashmir 181221, India
| | - Deepika Trakroo
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jammu, Jammu, Jammu and Kashmir 181221, India
| | - Neeraj Soni
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jammu, Jammu, Jammu and Kashmir 181221, India
| | - Punita Kumari
- Indian Institute of Science Education and Research Bhopal (IISERB), Department of Biological Sciences, Bhopal, Madhya Pradesh 462066, India
| | - Mithu Baidya
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jammu, Jammu, Jammu and Kashmir 181221, India
| |
Collapse
|
7
|
Chevigné A, Legler DF, Rot A, Sozzani S, Szpakowska M, Thelen M. International Union of Basic and Clinical Pharmacology. CXVIII. Update on the nomenclature for atypical chemokine receptors, including ACKR5. Pharmacol Rev 2025; 77:100012. [PMID: 39952689 DOI: 10.1124/pharmrev.124.001361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/26/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024] Open
Abstract
Chemokines signal through classical G protein-coupled receptors to induce cell migration during development, immune homeostasis, and multiple diseases. Over the last decade, a subfamily of atypical chemokine receptors (ACKRs) was delineated from G protein-coupled receptors based on their inability to trigger conventional G protein signaling or mediate cell migration in response to chemokines. These receptors nevertheless play an important role within the chemokine system by sequestering, transporting, or internalizing chemokines, thereby regulating their availability and shaping their gradients. GPR182, the recently deorphanized chemokine receptor, shares about 30% of sequence similarity with its closest relative ACKR3. GPR182 is mainly expressed on endothelial cells and was proposed to act as a scavenger regulating the availability of a large set of chemokines from the CXC, CC, and XC families and to act cooperatively with ACKR3 and ACKR4. Unlike other ACKRs, GPR182 was shown to have a strong constitutive interaction with β-arrestins that is required for intracellular receptor trafficking and chemokine scavenging. Chemokine ligation of GPR182 has no additional detectable impact on β-arrestin recruitment. Genetic ablation of GPR182 affects spleen size, myelopoiesis, and serum chemokine levels, indicating its role in chemokine homeostasis and immune regulation. GPR182 was also reported to regulate immune responses to bloodborne antigens and tumorigenesis. Taken together, compelling cumulative evidence indicates that GPR182 does not trigger G protein-mediated signaling but acts as a scavenger for chemokines in vitro and in vivo, strongly supporting its inclusion as ACKR5 in the systematic nomenclature of chemokine receptors. SIGNIFICANCE STATEMENT: The summarized presented findings strongly support the designation of GPR182 as ACKR5 and its formal inclusion in the family of ACKRs.
Collapse
Affiliation(s)
- Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Daniel F Legler
- Institute of Cell Biology and Immunology Thurgau (BITG), University of Konstanz, Kreuzlingen, Switzerland; Faculty of Biology, University of Konstanz, Konstanz, Germany; Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Antal Rot
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, Germany
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
8
|
Esmaeili AJ, Montazeri P, Gomez JC, Dumervil DJ, Nezhad FS, Steinhardt RC. Photoswitchable TCB-2 for control of the 5-HT 2A receptor and analysis of biased agonism. Chem Commun (Camb) 2024; 60:11956-11959. [PMID: 39350732 DOI: 10.1039/d4cc03892d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
Abstract
Therapies that target the serotonin 2A receptor (5-HT2AR) are promising. However, probes are needed to better understand the role of 5-HT2AR. Here, we design and synthesize a photoswitch and photoswitchable 5-HT2AR ligand based on highly potent agonist TCB-2 and arylazopyrazole, which also boasts photoswitchable G protein vs. β-arrestin pathway bias.
Collapse
Affiliation(s)
| | - Pantea Montazeri
- Syracuse University, Department of Chemistry, 111 College Pl., Syracuse, NY 13244, USA.
| | | | - Didier J Dumervil
- Syracuse University, Department of Chemistry, 111 College Pl., Syracuse, NY 13244, USA.
| | - Faezeh Safar Nezhad
- Syracuse University, Department of Chemistry, 111 College Pl., Syracuse, NY 13244, USA.
| | - Rachel C Steinhardt
- Syracuse University, Department of Chemistry, 111 College Pl., Syracuse, NY 13244, USA.
| |
Collapse
|
9
|
Tobin AB. A golden age of muscarinic acetylcholine receptor modulation in neurological diseases. Nat Rev Drug Discov 2024; 23:743-758. [PMID: 39143241 DOI: 10.1038/s41573-024-01007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/16/2024]
Abstract
Over the past 40 years, the muscarinic acetylcholine receptor family, particularly the M1-receptor and M4-receptor subtypes, have emerged as validated targets for the symptomatic treatment of neurological diseases such as schizophrenia and Alzheimer disease. However, despite considerable effort and investment, no drugs have yet gained clinical approval. This is largely attributable to cholinergic adverse effects that have halted the majority of programmes and resulted in a waning of interest in these G-protein-coupled receptor targets. Recently, this trend has been reversed. Driven by advances in structure-based drug design and an appreciation of the optimal pharmacological properties necessary to deliver clinical efficacy while minimizing adverse effects, a new generation of M1-receptor and M4-receptor orthosteric agonists and positive allosteric modulators are now entering the clinic. These agents offer the prospect of novel therapeutic solutions for 'hard to treat' neurological diseases, heralding a new era of muscarinic drug discovery.
Collapse
Affiliation(s)
- Andrew B Tobin
- Centre for Translational Pharmacology, School of Molecular Biosciences, The Advanced Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
10
|
Dai HC, Ji RL, Tao YX. SHU9119 and MBP10 are biased ligands at the human melanocortin-4 receptor. Biochem Pharmacol 2024; 228:116325. [PMID: 38815629 DOI: 10.1016/j.bcp.2024.116325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The melanocortin-4 receptor (MC4R), a G protein-coupled receptor, is critically involved in regulating energy homeostasis as well as modulation of reproduction and sexual function. Two peptide antagonists (SHU9119 and MBP10) were derived from the endogenous agonist α-melanocyte stimulating hormone. But their pharmacology at human MC4R is not fully understood. Herein, we performed detailed pharmacological studies of SHU9119 and MBP10 on wild-type (WT) and six naturally occurring constitutively active MC4Rs. Both ligands had no or negligible agonist activity in Gαs-cAMP signaling on WT MC4R, but stimulated extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation on WT and mutant MC4Rs. Mechanistic studies revealed that SHU9119 and MBP10 stimulated ERK1/2 signaling of MC4R by different mechanisms, with SHU9119-stimulated ERK1/2 signaling mediated by phosphatidylinositol 3-kinase (PI3K) and MBP10-initiated ERK1/2 activation through PI3K and β-arrestin. In summary, our studies demonstrated that SHU9119 and MBP10 were biased ligands for MC4R, preferentially activating ERK1/2 signaling through different mechanisms. SHU9119 acted as a biased ligand and MBP10 behaved as a biased allosteric modulator.
Collapse
Affiliation(s)
- Han-Chuan Dai
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| | - Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
11
|
Kogut-Günthel MM, Zara Z, Nicoli A, Steuer A, Lopez-Balastegui M, Selent J, Karanth S, Koehler M, Ciancetta A, Abiko LA, Hagn F, Di Pizio A. The path to the G protein-coupled receptor structural landscape: Major milestones and future directions. Br J Pharmacol 2024. [PMID: 39209310 DOI: 10.1111/bph.17314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/14/2024] [Accepted: 06/28/2024] [Indexed: 09/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) play a crucial role in cell function by transducing signals from the extracellular environment to the inside of the cell. They mediate the effects of various stimuli, including hormones, neurotransmitters, ions, photons, food tastants and odorants, and are renowned drug targets. Advancements in structural biology techniques, including X-ray crystallography and cryo-electron microscopy (cryo-EM), have driven the elucidation of an increasing number of GPCR structures. These structures reveal novel features that shed light on receptor activation, dimerization and oligomerization, dichotomy between orthosteric and allosteric modulation, and the intricate interactions underlying signal transduction, providing insights into diverse ligand-binding modes and signalling pathways. However, a substantial portion of the GPCR repertoire and their activation states remain structurally unexplored. Future efforts should prioritize capturing the full structural diversity of GPCRs across multiple dimensions. To do so, the integration of structural biology with biophysical and computational techniques will be essential. We describe in this review the progress of nuclear magnetic resonance (NMR) to examine GPCR plasticity and conformational dynamics, of atomic force microscopy (AFM) to explore the spatial-temporal dynamics and kinetic aspects of GPCRs, and the recent breakthroughs in artificial intelligence for protein structure prediction to characterize the structures of the entire GPCRome. In summary, the journey through GPCR structural biology provided in this review illustrates how far we have come in decoding these essential proteins architecture and function. Looking ahead, integrating cutting-edge biophysics and computational tools offers a path to navigating the GPCR structural landscape, ultimately advancing GPCR-based applications.
Collapse
Affiliation(s)
| | - Zeenat Zara
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Faculty of Science, University of South Bohemia in Ceske Budejovice, České Budějovice, Czech Republic
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Professorship for Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Alexandra Steuer
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Professorship for Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Marta Lopez-Balastegui
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute & Pompeu Fabra University, Barcelona, Spain
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute & Pompeu Fabra University, Barcelona, Spain
| | - Sanjai Karanth
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Melanie Koehler
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- TUM Junior Fellow at the Chair of Nutritional Systems Biology, Technical University of Munich, Freising, Germany
| | - Antonella Ciancetta
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Layara Akemi Abiko
- Focal Area Structural Biology and Biophysics, Biozentrum, University of Basel, Basel, Switzerland
| | - Franz Hagn
- Structural Membrane Biochemistry, Bavarian NMR Center, Dept. Bioscience, School of Natural Sciences, Technical University of Munich, Munich, Germany
- Institute of Structural Biology (STB), Helmholtz Munich, Neuherberg, Germany
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Professorship for Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| |
Collapse
|
12
|
Yuan Y, Xu T, Huang Y, Shi J. Strategies for developing μ opioid receptor agonists with reduced adverse effects. Bioorg Chem 2024; 149:107507. [PMID: 38850778 DOI: 10.1016/j.bioorg.2024.107507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/02/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Opioids are currently the most effective and widely used painkillers in the world. Unfortunately, the clinical use of opioid analgesics is limited by serious adverse effects. Many researchers have been working on designing and optimizing structures in search of novel μ opioid receptor(MOR) agonists with improved analgesic activity and reduced incidence of adverse effects. There are many strategies to develop MOR drugs, mainly focusing on new low efficacy agonists (potentially G protein biased agonists), MOR agonists acting on different Gα subtype, targeting opioid receptors in the periphery, acting on multiple opioid receptor, and targeting allosteric sites of opioid receptors, and others. This review summarizes the design methods, clinical applications, and structure-activity relationships of small-molecule agonists for MOR based on these different design strategies, providing ideas for the development of safer novel opioid ligands with therapeutic potential.
Collapse
Affiliation(s)
- Yan Yuan
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 611756, China
| | - Ting Xu
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Yu Huang
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 611756, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
13
|
Ünsal Ö, Bacaksiz ZS, Khamraev V, Montanari V, Beinborn M, Kumar K. Prolonged Activation of the GLP-1 Receptor via Covalent Capture. ACS Chem Biol 2024; 19:1453-1465. [PMID: 38935975 DOI: 10.1021/acschembio.4c00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
The incretin gut hormone glucagon-like peptide-1 (GLP-1) has become a household name because of its ability to induce glucose-dependent insulin release with accompanying weight loss in patients. Indeed, derivatives of the peptide exert numerous pleiotropic actions that favorably affect other metabolic functions, and consequently, such compounds are being considered as treatments for a variety of ailments. The ability of native GLP-1 to function as a clinical drug is severely limited because of its short half-life in vivo. All of the beneficial effects of GLP-1 come from its agonism at the cognate receptor, GLP-1R. In our quest for long-lived activation of the receptor, we hypothesized that an agonist that had the ability to covalently cross-link with GLP-1R would prove useful. We here report the structure-guided design of peptide analogues containing an electrophilic warhead that could be covalently captured by a resident native nucleophile on the receptor. The compounds were evaluated using washout experiments, and resistance to such washing serves as an index of prolonged activation and covalent capture, which we use to tabulate longevity and robust long-lived GLP-1R agonism. The addition of SulF (cross-linkable warhead), an N-terminal trifluoroethyl group (for protease protection), and a C18 diacid lipid (protractor) all contributed to the increased wash resistance of GLP-1. The most effective compound based on the wash resistance metric, C2K26DAC18_K34SulF, has all three elements outlined and may serve as a blueprint and a proof-of-concept scaffold for the design of clinically useful molecules.
Collapse
Affiliation(s)
- Özge Ünsal
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Z Selin Bacaksiz
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Vladislav Khamraev
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Vittorio Montanari
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Martin Beinborn
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
- Molecular Pharmacology Research Center, Tufts Medical Center, Boston, Massachusetts 02111, United States
| | - Krishna Kumar
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
14
|
Yu F, Zong B, Ji L, Sun P, Jia D, Wang R. Free Fatty Acids and Free Fatty Acid Receptors: Role in Regulating Arterial Function. Int J Mol Sci 2024; 25:7853. [PMID: 39063095 PMCID: PMC11277118 DOI: 10.3390/ijms25147853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The metabolic network's primary sources of free fatty acids (FFAs) are long- and medium-chain fatty acids of triglyceride origin and short-chain fatty acids produced by intestinal microorganisms through dietary fibre fermentation. Recent studies have demonstrated that FFAs not only serve as an energy source for the body's metabolism but also participate in regulating arterial function. Excess FFAs have been shown to lead to endothelial dysfunction, vascular hypertrophy, and vessel wall stiffness, which are important triggers of arterial hypertension and atherosclerosis. Nevertheless, free fatty acid receptors (FFARs) are involved in the regulation of arterial functions, including the proliferation, differentiation, migration, apoptosis, inflammation, and angiogenesis of vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs). They actively regulate hypertension, endothelial dysfunction, and atherosclerosis. The objective of this review is to examine the roles and heterogeneity of FFAs and FFARs in the regulation of arterial function, with a view to identifying the points of intersection between their actions and providing new insights into the prevention and treatment of diseases associated with arterial dysfunction, as well as the development of targeted drugs.
Collapse
Affiliation(s)
- Fengzhi Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (B.Z.); (P.S.)
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lili Ji
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (B.Z.); (P.S.)
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (F.Y.); (L.J.)
| |
Collapse
|
15
|
Chesnokov MS, Mamedova AR, Zhivotovsky B, Kopeina GS. A matter of new life and cell death: programmed cell death in the mammalian ovary. J Biomed Sci 2024; 31:31. [PMID: 38509545 PMCID: PMC10956231 DOI: 10.1186/s12929-024-01017-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND The mammalian ovary is a unique organ that displays a distinctive feature of cyclic changes throughout the entire reproductive period. The estrous/menstrual cycles are associated with drastic functional and morphological rearrangements of ovarian tissue, including follicular development and degeneration, and the formation and subsequent atrophy of the corpus luteum. The flawless execution of these reiterative processes is impossible without the involvement of programmed cell death (PCD). MAIN TEXT PCD is crucial for efficient and careful clearance of excessive, depleted, or obsolete ovarian structures for ovarian cycling. Moreover, PCD facilitates selection of high-quality oocytes and formation of the ovarian reserve during embryonic and juvenile development. Disruption of PCD regulation can heavily impact the ovarian functions and is associated with various pathologies, from a moderate decrease in fertility to severe hormonal disturbance, complete loss of reproductive function, and tumorigenesis. This comprehensive review aims to provide updated information on the role of PCD in various processes occurring in normal and pathologic ovaries. Three major events of PCD in the ovary-progenitor germ cell depletion, follicular atresia, and corpus luteum degradation-are described, alongside the detailed information on molecular regulation of these processes, highlighting the contribution of apoptosis, autophagy, necroptosis, and ferroptosis. Ultimately, the current knowledge of PCD aberrations associated with pathologies, such as polycystic ovarian syndrome, premature ovarian insufficiency, and tumors of ovarian origin, is outlined. CONCLUSION PCD is an essential element in ovarian development, functions and pathologies. A thorough understanding of molecular mechanisms regulating PCD events is required for future advances in the diagnosis and management of various disorders of the ovary and the female reproductive system in general.
Collapse
Affiliation(s)
- Mikhail S Chesnokov
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Aygun R Mamedova
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Boris Zhivotovsky
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Gelina S Kopeina
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
16
|
Michinaga S, Nagata A, Ogami R, Ogawa Y, Hishinuma S. Histamine H 1 Receptor-Mediated JNK Phosphorylation Is Regulated by G q Protein-Dependent but Arrestin-Independent Pathways. Int J Mol Sci 2024; 25:3395. [PMID: 38542369 PMCID: PMC10970263 DOI: 10.3390/ijms25063395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Arrestins are known to be involved not only in the desensitization and internalization of G protein-coupled receptors but also in the G protein-independent activation of mitogen-activated protein (MAP) kinases, such as extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), to regulate cell proliferation and inflammation. Our previous study revealed that the histamine H1 receptor-mediated activation of ERK is dually regulated by Gq proteins and arrestins. In this study, we investigated the roles of Gq proteins and arrestins in the H1 receptor-mediated activation of JNK in Chinese hamster ovary (CHO) cells expressing wild-type (WT) human H1 receptors, the Gq protein-biased mutant S487TR, and the arrestin-biased mutant S487A. In these mutants, the Ser487 residue in the C-terminus region of the WT was truncated (S487TR) or mutated to alanine (S487A). Histamine significantly stimulated JNK phosphorylation in CHO cells expressing WT and S487TR but not S487A. Histamine-induced JNK phosphorylation in CHO cells expressing WT and S487TR was suppressed by inhibitors against H1 receptors (ketotifen and diphenhydramine), Gq proteins (YM-254890), and protein kinase C (PKC) (GF109203X) as well as an intracellular Ca2+ chelator (BAPTA-AM) but not by inhibitors against G protein-coupled receptor kinases (GRK2/3) (cmpd101), β-arrestin2 (β-arrestin2 siRNA), and clathrin (hypertonic sucrose). These results suggest that the H1 receptor-mediated phosphorylation of JNK is regulated by Gq-protein/Ca2+/PKC-dependent but GRK/arrestin/clathrin-independent pathways.
Collapse
Affiliation(s)
| | | | | | | | - Shigeru Hishinuma
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan
| |
Collapse
|
17
|
Kee TR, Khan SA, Neidhart MB, Masters BM, Zhao VK, Kim YK, McGill Percy KC, Woo JAA. The multifaceted functions of β-arrestins and their therapeutic potential in neurodegenerative diseases. Exp Mol Med 2024; 56:129-141. [PMID: 38212557 PMCID: PMC10834518 DOI: 10.1038/s12276-023-01144-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 01/13/2024] Open
Abstract
Arrestins are multifunctional proteins that regulate G-protein-coupled receptor (GPCR) desensitization, signaling, and internalization. The arrestin family consists of four subtypes: visual arrestin1, β-arrestin1, β-arrestin2, and visual arrestin-4. Recent studies have revealed the multifunctional roles of β-arrestins beyond GPCR signaling, including scaffolding and adapter functions, and physically interacting with non-GPCR receptors. Increasing evidence suggests that β-arrestins are involved in the pathogenesis of a variety of neurodegenerative diseases, including Alzheimer's disease (AD), frontotemporal dementia (FTD), and Parkinson's disease (PD). β-arrestins physically interact with γ-secretase, leading to increased production and accumulation of amyloid-beta in AD. Furthermore, β-arrestin oligomers inhibit the autophagy cargo receptor p62/SQSTM1, resulting in tau accumulation and aggregation in FTD. In PD, β-arrestins are upregulated in postmortem brain tissue and an MPTP model, and the β2AR regulates SNCA gene expression. In this review, we aim to provide an overview of β-arrestin1 and β-arrestin2, and describe their physiological functions and roles in neurodegenerative diseases. The multifaceted roles of β-arrestins and their involvement in neurodegenerative diseases suggest that they may serve as promising therapeutic targets.
Collapse
Affiliation(s)
- Teresa R Kee
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
- Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, 33613, USA
| | - Sophia A Khan
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Maya B Neidhart
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Brianna M Masters
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Victoria K Zhao
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Yenna K Kim
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | | | - Jung-A A Woo
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
18
|
Madhu MK, Shewani K, Murarka RK. Biased Signaling in Mutated Variants of β 2-Adrenergic Receptor: Insights from Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:449-469. [PMID: 38194225 DOI: 10.1021/acs.jcim.3c01481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
The molecular basis of receptor bias in G protein-coupled receptors (GPCRs) caused by mutations that preferentially activate specific intracellular transducers over others remains poorly understood. Two experimentally identified biased variants of β2-adrenergic receptors (β2AR), a prototypical GPCR, are a triple mutant (T68F, Y132A, and Y219A) and a single mutant (Y219A); the former bias the receptor toward the β-arrestin pathway by disfavoring G protein engagement, while the latter induces G protein signaling explicitly due to selection against GPCR kinases (GRKs) that phosphorylate the receptor as a prerequisite of β-arrestin binding. Though rigorous characterizations have revealed functional implications of these mutations, the atomistic origin of the observed transducer selectivity is not clear. In this study, we investigated the allosteric mechanism of receptor bias in β2AR using microseconds of all-atom Gaussian accelerated molecular dynamics (GaMD) simulations. Our observations reveal distinct rearrangements in transmembrane helices, intracellular loop 3, and critical residues R1313.50 and Y3267.53 in the conserved motifs D(E)RY and NPxxY for the mutant receptors, leading to their specific transducer interactions. Moreover, partial dissociation of G protein from the receptor core is observed in the simulations of the triple mutant in contrast to the single mutant and wild-type receptor. The reorganization of allosteric communications from the extracellular agonist BI-167107 to the intracellular receptor-transducer interfaces drives the conformational rearrangements responsible for receptor bias in the single and triple mutants. The molecular insights into receptor bias of β2AR presented here could improve the understanding of biased signaling in GPCRs, potentially opening new avenues for designing novel therapeutics with fewer side-effects and superior efficacy.
Collapse
Affiliation(s)
- Midhun K Madhu
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh 462066, India
| | - Kunal Shewani
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh 462066, India
| | - Rajesh K Murarka
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh 462066, India
| |
Collapse
|
19
|
Maharana J, Sano FK, Sarma P, Yadav MK, Duan L, Stepniewski TM, Chaturvedi M, Ranjan A, Singh V, Saha S, Mahajan G, Chami M, Shihoya W, Selent J, Chung KY, Banerjee R, Nureki O, Shukla AK. Molecular insights into atypical modes of β-arrestin interaction with seven transmembrane receptors. Science 2024; 383:101-108. [PMID: 38175886 PMCID: PMC7615931 DOI: 10.1126/science.adj3347] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
β-arrestins (βarrs) are multifunctional proteins involved in signaling and regulation of seven transmembrane receptors (7TMRs), and their interaction is driven primarily by agonist-induced receptor activation and phosphorylation. Here, we present seven cryo-electron microscopy structures of βarrs either in the basal state, activated by the muscarinic receptor subtype 2 (M2R) through its third intracellular loop, or activated by the βarr-biased decoy D6 receptor (D6R). Combined with biochemical, cellular, and biophysical experiments, these structural snapshots allow the visualization of atypical engagement of βarrs with 7TMRs and also reveal a structural transition in the carboxyl terminus of βarr2 from a β strand to an α helix upon activation by D6R. Our study provides previously unanticipated molecular insights into the structural and functional diversity encoded in 7TMR-βarr complexes with direct implications for exploring novel therapeutic avenues.
Collapse
Affiliation(s)
- Jagannath Maharana
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Fumiya K. Sano
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Parishmita Sarma
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Manish K. Yadav
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Longhan Duan
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tomasz M. Stepniewski
- Research Program on Biomedical Informatics, Hospital del Mar Research Institute and Pompeu Fabra University, Barcelona, Spain
| | - Madhu Chaturvedi
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Ashutosh Ranjan
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Vinay Singh
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Sayantan Saha
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Gargi Mahajan
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Mohamed Chami
- BioEM Lab, Biozentrum, University of Basel, Basel, Switzerland
| | - Wataru Shihoya
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Jana Selent
- Research Program on Biomedical Informatics, Hospital del Mar Research Institute and Pompeu Fabra University, Barcelona, Spain
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ramanuj Banerjee
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| | - Osamu Nureki
- Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Arun K. Shukla
- Department of Biological Sciences, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
20
|
Wang S, Peng L, Kim KM. Biased Dopamine D 2 Receptors Exhibit Distinct Intracellular Trafficking Properties and ERK Activation in Different Subcellular Domains. Biomol Ther (Seoul) 2024; 32:56-64. [PMID: 37465849 PMCID: PMC10762269 DOI: 10.4062/biomolther.2023.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 06/18/2023] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
Biased signaling or functional selectivity refers to the ability of an agonist or receptor to selectively activate a subset of transducers such as G protein and arrestin in the case of G protein-coupled receptors (GPCRs). Although signaling through arrestin has been reported from various GPCRs, only a few studies have examined side-by-side how it differs from signaling via G protein. In this study, two signaling pathways were compared using dopamine D2 receptor (D2R) mutants engineered via the evolutionary tracer method to selectively transduce signals through G protein or arrestin (D2G and D2Arr, respectively). D2G mediated the inhibition of cAMP production and ERK activation in the cytoplasm. D2Arr, in contrast, mediated receptor endocytosis accompanied by arrestin ubiquitination and ERK activation in the nucleus as well as in the cytoplasm. D2Arr-mediated ERK activation occurred in a manner dependent on arrestin3 but not arrestin2, accompanied by the nuclear translocation of arrestin3 via importin1. D2R-mediated ERK activation, which occurred in both the cytosol and nucleus, was limited to the cytosol when cellular arrestin3 was depleted. This finding supports the results obtained with D2Arr and D2G. Taken together, these observations indicate that biased signal transduction pathways activate distinct downstream mechanisms and that the subcellular regions in which they occur could be different when the same effectors are involved. These findings broaden our understanding on the relation between biased receptors and the corresponding downstream signaling, which is critical for elucidating the functional roles of biased pathways.
Collapse
Affiliation(s)
- Shujie Wang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Lulu Peng
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
21
|
Naser NH, Suhail FSA, Hussein SA, Salih SS. Physicochemical properties as a function of lomefloxacin biological activity. POLSKI MERKURIUSZ LEKARSKI : ORGAN POLSKIEGO TOWARZYSTWA LEKARSKIEGO 2024; 52:197-202. [PMID: 38642355 DOI: 10.36740/merkur202402108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2024]
Abstract
OBJECTIVE Aim: The goal is to discover QSAR of Lomefloxacin as antibacterial activity. PATIENTS AND METHODS Materials and Methods: A number of lomefloxacins analogs activities were studied by program Windows Chem SW. The analogues were obtained and energy minimization was carried out through Molecular Modeling Program, the calculations were performed using General Atomic and Molecular Electronic Structure System (GAMESS) software. RESULTS Results: There were six descriptions (N-quinoline more (-) ev charge, Kinetic Energy, Potential Energy, Log p, Log S, F6 charge) results have highly compatible of physicochemical properties with lomefloxacin analogs activities. It can be used to estimate the activities depending on QSAR equation of lomefloxacin analogs. CONCLUSION Conclusions: The parameters used for calculation were depending on the quantum chemical was employed in deriving from computational study of properties and can used to predict the activities of certain analogs of Lomefloxacins as antibacterial compounds.
Collapse
|
22
|
Eissa AM, Hassanin MH, Ibrahim IAAEH. Hepatic β-arrestins: potential roles in liver health and disease. Mol Biol Rep 2023; 50:10399-10407. [PMID: 37843713 PMCID: PMC10676313 DOI: 10.1007/s11033-023-08898-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023]
Abstract
Β-arrestins are intracellular scaffolding proteins that have multifaceted roles in different types of disorders. In this review article, we gave a summary about the discovery, characterization and classification of these proteins and their intracellular functions. Moreover, this review article focused on the hepatic expression of β-arrestins and their hepatocellular distribution and function in each liver cell type. Also, we showed that β-arrestins are key regulators of distinct types of hepatic disorders. On the other hand, we addressed some important points that have never been studied before regarding the role of β-arrestins in certain types of hepatic disorders which needs more research efforts to cover.
Collapse
Affiliation(s)
| | | | - Islam A A E H Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
23
|
Zehnaker A, Vallet A, Gourdon J, Sarti C, Jugnarain V, Haj Hassan M, Mathias L, Gauthier C, Raynaud P, Boulo T, Beauclair L, Bigot Y, Casarini L, Crépieux P, Poupon A, Piégu B, Jean-Alphonse F, Bruneau G, Reiter É. Combined Multiplexed Phage Display, High-Throughput Sequencing, and Functional Assays as a Platform for Identifying Modulatory VHHs Targeting the FSHR. Int J Mol Sci 2023; 24:15961. [PMID: 37958944 PMCID: PMC10650796 DOI: 10.3390/ijms242115961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Developing modulatory antibodies against G protein-coupled receptors is challenging. In this study, we targeted the follicle-stimulating hormone receptor (FSHR), a significant regulator of reproduction, with variable domains of heavy chain-only antibodies (VHHs). We built two immune VHH libraries and submitted them to multiplexed phage display approaches. We used next-generation sequencing to identify 34 clusters of specifically enriched sequences that were functionally assessed in a primary screen based on a cAMP response element (CRE)-dependent reporter gene assay. In this assay, 23 VHHs displayed negative or positive modulation of FSH-induced responses, suggesting a high success rate of the multiplexed strategy. We then focused on the largest cluster identified (i.e., PRC1) that displayed positive modulation of FSH action. We demonstrated that PRC1 specifically binds to the human FSHR and human FSHR/FSH complex while potentiating FSH-induced cAMP production and Gs recruitment. We conclude that the improved selection strategy reported here is effective for rapidly identifying functionally active VHHs and could be adapted to target other challenging membrane receptors. This study also led to the identification of PRC1, the first potential positive modulator VHH reported for the human FSHR.
Collapse
Affiliation(s)
- Anielka Zehnaker
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Amandine Vallet
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Juliette Gourdon
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Caterina Sarti
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Vinesh Jugnarain
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Maya Haj Hassan
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Laetitia Mathias
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Camille Gauthier
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Pauline Raynaud
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Thomas Boulo
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Linda Beauclair
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Yves Bigot
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Livio Casarini
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Pascale Crépieux
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| | - Anne Poupon
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
- MAbSilico, 1 Impasse du Palais, 37000 Tours, France
| | - Benoît Piégu
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Frédéric Jean-Alphonse
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| | - Gilles Bruneau
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Éric Reiter
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| |
Collapse
|
24
|
Gutierrez Cruz A, Aresta Branco MSL, Borhani Peikani M, Mutafova-Yambolieva VN. Differential Influences of Endogenous and Exogenous Sensory Neuropeptides on the ATP Metabolism by Soluble Ectonucleotidases in the Murine Bladder Lamina Propria. Int J Mol Sci 2023; 24:15650. [PMID: 37958631 PMCID: PMC10647406 DOI: 10.3390/ijms242115650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Bladder urothelium and suburothelium/lamina propria (LP) have prominent sensory and transducer functions with the active participation of afferent neurons and urothelium-derived purine mediators such as adenosine 5'-triphosphate (ATP), adenosine 5'-diphosphate (ADP), and adenosine (ADO). Effective concentrations of purines at receptor targets depend significantly on the extracellular degradation of ATP by ectonucleotidases (ENTDs). We recently reported the regulated release of soluble ENTDs (s-ENTDs) in the LP and the consequent degradation of ATP to ADP, AMP, and ADO. Afferent neurons in the LP can be activated by urothelial ATP and release peptides and other transmitters that can alter the activity of cells in their vicinity. Using a murine decentralized ex vivo detrusor-free bladder model, 1,N6-etheno-ATP (eATP) as substrate, and sensitive HPLC-FLD methodologies, we found that exogenous neuropeptides calcitonin gene-related peptide (CGRP), substance P (Sub P), neurokinin A (NKA), and pituitary adenylate cyclase-activating polypeptide [PACAP (1-38)] all increased the degradation of eATP by s-ENTDs that were released in the LP spontaneously and/or during bladder filling. Using antagonists of neuropeptide receptors, we observed that endogenous NKA did not modify the ATP hydrolysis by s-ENTDs, whereas endogenous Sub P increased both the constitutive and distention-induced release of s-ENTDs. In contrast, endogenous CGRP and PACAP (1-38) increased the distention-induced, but not the spontaneous, release of s-ENTDs. The present study puts forward the novel idea that interactions between peptidergic and purinergic signaling mechanisms in the LP have an impact on bladder excitability and functions by regulating the effective concentrations of adenine purines at effector cells in the LP.
Collapse
Affiliation(s)
| | | | | | - Violeta N. Mutafova-Yambolieva
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV 89557, USA; (A.G.C.); (M.B.P.)
| |
Collapse
|
25
|
Matt RA, Westhorpe FG, Romuar RF, Rana P, Gever JR, Ford AP. Fingerprinting heterocellular β-adrenoceptor functional expression in the brain using agonist activity profiles. Front Mol Biosci 2023; 10:1214102. [PMID: 37664183 PMCID: PMC10471193 DOI: 10.3389/fmolb.2023.1214102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/05/2023] [Indexed: 09/05/2023] Open
Abstract
Noradrenergic projections from the brainstem locus coeruleus drive arousal, attentiveness, mood, and memory, but specific adrenoceptor (AR) function across the varied brain cell types has not been extensively characterized, especially with agonists. This study reports a pharmacological analysis of brain AR function, offering insights for innovative therapeutic interventions that might serve to compensate for locus coeruleus decline, known to develop in the earliest phases of neurodegenerative diseases. First, β-AR agonist activities were measured in recombinant cell systems and compared with those of isoprenaline to generate Δlog(Emax/EC50) values, system-independent metrics of agonist activity, that, in turn, provide receptor subtype fingerprints. These fingerprints were then used to assess receptor subtype expression across human brain cell systems and compared with Δlog(Emax/EC50) values arising from β-arrestin activation or measurements of cAMP response desensitization to assess the possibility of ligand bias among β-AR agonists. Agonist activity profiles were confirmed to be system-independent and, in particular, revealed β2-AR functional expression across several human brain cell types. Broad β2-AR function observed is consistent with noradrenergic tone arising from the locus coeruleus exerting heterocellular neuroexcitatory and homeostatic influence. Notably, Δlog(Emax/EC50) measurements suggest that tested β-AR agonists do not show ligand bias as it pertains to homologous receptor desensitization in the system examined. Δlog(Emax/EC50) agonist fingerprinting is a powerful means of assessing receptor subtype expression regardless of receptor expression levels or assay readout, and the method may be applicable to future use for novel ligands and tissues expressing any receptor with available reference agonists.
Collapse
|
26
|
Ji RL, Liu T, Hou ZS, Wen HS, Tao YX. Divergent Pharmacology and Biased Signaling of the Four Melanocortin-4 Receptor Isoforms in Rainbow Trout ( Oncorhynchus mykiss). Biomolecules 2023; 13:1248. [PMID: 37627313 PMCID: PMC10452266 DOI: 10.3390/biom13081248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
The melanocortin-4 receptor (MC4R) is essential for the modulation of energy balance and reproduction in both fish and mammals. Rainbow trout (Oncorhynchus mykiss) has been extensively studied in various fields and provides a unique opportunity to investigate divergent physiological roles of paralogues. Herein we identified four trout mc4r (mc4ra1, mc4ra2, mc4rb1, and mc4rb2) genes. Four trout Mc4rs (omMc4rs) were homologous to those of teleost and mammalian MC4Rs. Multiple sequence alignments, a phylogenetic tree, chromosomal synteny analyses, and pharmacological studies showed that trout mc4r genes may have undergone different evolutionary processes. All four trout Mc4rs bound to two peptide agonists and elevated intracellular cAMP levels dose-dependently. High basal cAMP levels were observed at two omMc4rs, which were decreased by Agouti-related peptide. Only omMc4rb2 was constitutively active in the ERK1/2 signaling pathway. Ipsen 5i, ML00253764, and MCL0020 were biased allosteric modulators of omMc4rb1 with selective activation upon ERK1/2 signaling. ML00253764 behaved as an allosteric agonist in Gs-cAMP signaling of omMc4rb2. This study will lay the foundation for future physiological studies of various mc4r paralogs and reveal the evolution of MC4R in vertebrates.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| | - Ting Liu
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| | - Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| | - Hai-Shen Wen
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266100, China;
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (R.-L.J.); (T.L.)
| |
Collapse
|
27
|
Singhal SM, Zell V, Faget L, Slosky LM, Barak LS, Caron MG, Pinkerton AB, Hnasko TS. Neurotensin receptor 1-biased ligand attenuates neurotensin-mediated excitation of ventral tegmental area dopamine neurons and dopamine release in the nucleus accumbens. Neuropharmacology 2023; 234:109544. [PMID: 37055008 PMCID: PMC10192038 DOI: 10.1016/j.neuropharm.2023.109544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
Strong expression of the G protein-coupled receptor (GPCR) neurotensin receptor 1 (NTR1) in ventral tegmental area (VTA) dopamine (DA) neurons and terminals makes it an attractive target to modulate DA neuron activity and normalize DA-related pathologies. Recent studies have identified a novel class of NTR1 ligand that shows promising effects in preclinical models of addiction. A lead molecule, SBI-0654553 (SBI-553), can act as a positive allosteric modulator of NTR1 β-arrestin recruitment while simultaneously antagonizing NTR1 Gq protein signaling. Using cell-attached recordings from mouse VTA DA neurons we discovered that, unlike neurotensin (NT), SBI-553 did not independently increase spontaneous firing. Instead, SBI-553 blocked the NT-mediated increase in firing. SBI-553 also antagonized the effects of NT on dopamine D2 auto-receptor signaling, potentially through its inhibitory effects on G-protein signaling. We also measured DA release directly, using fast-scan cyclic voltammetry in the nucleus accumbens and observed antagonist effects of SBI-553 on an NT-induced increase in DA release. Further, in vivo administration of SBI-553 did not notably change basal or cocaine-evoked DA release measured in NAc using fiber photometry. Overall, these results indicate that SBI-553 blunts NT's effects on spontaneous DA neuron firing, D2 auto-receptor function, and DA release, without independently affecting these measures. In the presence of NT, SBI-553 has an inhibitory effect on mesolimbic DA activity, which could contribute to its efficacy in animal models of psychostimulant use.
Collapse
Affiliation(s)
- Sarthak M Singhal
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Vivien Zell
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Lauren Faget
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Lauren M Slosky
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | | | - Marc G Caron
- Departments of Cell Biology, Neurobiology and Medicine, Duke University, Durham, NC, USA
| | - Anthony B Pinkerton
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Thomas S Hnasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
28
|
Sarma P, Carino CMC, Seetharama D, Pandey S, Dwivedi-Agnihotri H, Rui X, Cao Y, Kawakami K, Kumari P, Chen YC, Luker KE, Yadav PN, Luker GD, Laporte SA, Chen X, Inoue A, Shukla AK. Molecular insights into intrinsic transducer-coupling bias in the CXCR4-CXCR7 system. Nat Commun 2023; 14:4808. [PMID: 37558722 PMCID: PMC10412580 DOI: 10.1038/s41467-023-40482-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
Chemokine receptors constitute an important subfamily of G protein-coupled receptors (GPCRs), and they are critically involved in a broad range of immune response mechanisms. Ligand promiscuity among these receptors makes them an interesting target to explore multiple aspects of biased agonism. Here, we comprehensively characterize two chemokine receptors namely, CXCR4 and CXCR7, in terms of their transducer-coupling and downstream signaling upon their stimulation by a common chemokine agonist, CXCL12, and a small molecule agonist, VUF11207. We observe that CXCR7 lacks G-protein-coupling while maintaining robust βarr recruitment with a major contribution of GRK5/6. On the other hand, CXCR4 displays robust G-protein activation as expected but exhibits significantly reduced βarr-coupling compared to CXCR7. These two receptors induce distinct βarr conformations even when activated by the same agonist, and CXCR7, unlike CXCR4, fails to activate ERK1/2 MAP kinase. We also identify a key contribution of a single phosphorylation site in CXCR7 for βarr recruitment and endosomal localization. Our study provides molecular insights into intrinsic-bias encoded in the CXCR4-CXCR7 system with broad implications for drug discovery.
Collapse
Affiliation(s)
- Parishmita Sarma
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Carlo Marion C Carino
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Deeksha Seetharama
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Shubhi Pandey
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Hemlata Dwivedi-Agnihotri
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Xue Rui
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Yubo Cao
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Poonam Kumari
- Neuroscience and Ageing Biology Division, CSIR-Central Drug Research Institute Sector 10, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Yu-Chih Chen
- Department of Computational and Systems Biology, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Prem N Yadav
- Neuroscience and Ageing Biology Division, CSIR-Central Drug Research Institute Sector 10, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Stéphane A Laporte
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
- Department of Medicine, McGill University Health Center, McGill University, Montréal, QC, H4A 3J1, Canada
| | - Xin Chen
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India.
| |
Collapse
|
29
|
Chen K, Zhang C, Lin S, Yan X, Cai H, Yi C, Ma L, Chu X, Liu Y, Zhu Y, Han S, Zhao Q, Wu B. Tail engagement of arrestin at the glucagon receptor. Nature 2023; 620:904-910. [PMID: 37558880 PMCID: PMC10447241 DOI: 10.1038/s41586-023-06420-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023]
Abstract
Arrestins have pivotal roles in regulating G protein-coupled receptor (GPCR) signalling by desensitizing G protein activation and mediating receptor internalization1,2. It has been proposed that the arrestin binds to the receptor in two different conformations, 'tail' and 'core', which were suggested to govern distinct processes of receptor signalling and trafficking3,4. However, little structural information is available for the tail engagement of the arrestins. Here we report two structures of the glucagon receptor (GCGR) bound to β-arrestin 1 (βarr1) in glucagon-bound and ligand-free states. These structures reveal a receptor tail-engaged binding mode of βarr1 with many unique features, to our knowledge, not previously observed. Helix VIII, instead of the receptor core, has a major role in accommodating βarr1 by forming extensive interactions with the central crest of βarr1. The tail-binding pose is further defined by a close proximity between the βarr1 C-edge and the receptor helical bundle, and stabilized by a phosphoinositide derivative that bridges βarr1 with helices I and VIII of GCGR. Lacking any contact with the arrestin, the receptor core is in an inactive state and loosely binds to glucagon. Further functional studies suggest that the tail conformation of GCGR-βarr governs βarr recruitment at the plasma membrane and endocytosis of GCGR, and provides a molecular basis for the receptor forming a super-complex simultaneously with G protein and βarr to promote sustained signalling within endosomes. These findings extend our knowledge about the arrestin-mediated modulation of GPCR functionalities.
Collapse
Affiliation(s)
- Kun Chen
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chenhui Zhang
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuling Lin
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinyu Yan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Heng Cai
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Cuiying Yi
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Limin Ma
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaojing Chu
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yuchen Liu
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ya Zhu
- Lingang Laboratory, Shanghai, China
| | - Shuo Han
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Qiang Zhao
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China.
| | - Beili Wu
- State Key Laboratory of Drug Research, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
30
|
Labani N, Gbahou F, Noblet M, Masri B, Broussaud O, Liu J, Jockers R. Pistacia vera Extract Potentiates the Effect of Melatonin on Human Melatonin MT 1 and MT 2 Receptors with Functional Selectivity. Pharmaceutics 2023; 15:1845. [PMID: 37514032 PMCID: PMC10386454 DOI: 10.3390/pharmaceutics15071845] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Melatonin is a tryptophan derivative synthesized in plants and animals. In humans, melatonin acts on melatonin MT1 and MT2 receptors belonging to the G protein-coupled receptor (GPCR) family. Synthetic melatonin receptor agonists are prescribed for insomnia and depressive and circadian-related disorders. Here, we tested 25 commercial plant extracts, reported to have beneficial properties in sleep disorders and anxiety, using cellular assays (2─[125I]iodomelatonin binding, cAMP inhibition, ERK1/2 activation and β-arrestin2 recruitment) in mock-transfected and HEK293 cells expressing MT1 or MT2. Various melatonin receptor-dependent and -independent effects were observed. Extract 18 (Ex18) from Pistacia vera dried fruits stood out with very potent effects in melatonin receptor expressing cells. The high content of endogenous melatonin in Ex18 (5.28 ± 0.46 mg/g extract) is consistent with this observation. Ex18 contains an additional active principle that potentiates the effect of melatonin on Gi protein-dependent pathways but not on β-arrestin2 recruitment. Further active principles potentiating exogenous melatonin were detected in several extracts. In conclusion, we identified plant extracts with various effects in GPCR-based binding and signalling assays and identified high melatonin levels and a melatonin-potentiating activity in Pistacia vera dried fruit extracts that might be of therapeutic potential.
Collapse
Affiliation(s)
- Nedjma Labani
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Institut Cochin, CNRS, INSERM, University of Paris, F-75014 Paris, France
| | - Florence Gbahou
- Institut Cochin, CNRS, INSERM, University of Paris, F-75014 Paris, France
| | - Marc Noblet
- Science Hub, Sanofi Consumer Healthcare, F-75017 Paris, France
| | - Bernard Masri
- Institut Cochin, CNRS, INSERM, University of Paris, F-75014 Paris, France
| | | | - Jianfeng Liu
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ralf Jockers
- Institut Cochin, CNRS, INSERM, University of Paris, F-75014 Paris, France
| |
Collapse
|
31
|
van de Wetering R, Ewald A, Welsh S, Kornberger L, Williamson SE, McElroy BD, Butelman ER, Prisinzano TE, Kivell BM. The Kappa Opioid Receptor Agonist 16-Bromo Salvinorin A Has Anti-Cocaine Effects without Significant Effects on Locomotion, Food Reward, Learning and Memory, or Anxiety and Depressive-like Behaviors. Molecules 2023; 28:4848. [PMID: 37375403 PMCID: PMC10304272 DOI: 10.3390/molecules28124848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Kappa opioid receptor (KOR) agonists have preclinical antipsychostimulant effects; however, adverse side effects have limited their therapeutic development. In this preclinical study, conducted in Sprague Dawley rats, B6-SJL mice, and non-human primates (NHPs), we evaluated the G-protein-biased analogue of salvinorin A (SalA), 16-bromo salvinorin A (16-BrSalA), for its anticocaine effects, side effects, and activation of cellular signaling pathways. 16-BrSalA dose-dependently decreased the cocaine-primed reinstatement of drug-seeking behavior in a KOR-dependent manner. It also decreased cocaine-induced hyperactivity, but had no effect on responding for cocaine on a progressive ratio schedule. Compared to SalA, 16-BrSalA had an improved side effect profile, with no significant effects in the elevated plus maze, light-dark test, forced swim test, sucrose self-administration, or novel object recognition; however, it did exhibit conditioned aversive effects. 16-BrSalA increased dopamine transporter (DAT) activity in HEK-293 cells coexpressing DAT and KOR, as well as in rat nucleus accumbens and dorsal striatal tissue. 16-BrSalA also increased the early phase activation of extracellular-signal-regulated kinases 1 and 2, as well as p38 in a KOR-dependent manner. In NHPs, 16-BrSalA caused dose-dependent increases in the neuroendocrine biomarker prolactin, similar to other KOR agonists, at doses without robust sedative effects. These findings highlight that G-protein-biased structural analogues of SalA can have improved pharmacokinetic profiles and fewer side effects while maintaining their anticocaine effects.
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Amy Ewald
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Susan Welsh
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Lindsay Kornberger
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA (T.E.P.)
| | - Samuel E. Williamson
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bryan D. McElroy
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Eduardo R. Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Thomas E. Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA (T.E.P.)
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bronwyn M. Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
32
|
Maharana J, Sarma P, Yadav MK, Saha S, Singh V, Saha S, Chami M, Banerjee R, Shukla AK. Structural snapshots uncover a key phosphorylation motif in GPCRs driving β-arrestin activation. Mol Cell 2023; 83:2091-2107.e7. [PMID: 37209686 PMCID: PMC7615930 DOI: 10.1016/j.molcel.2023.04.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/22/2023] [Accepted: 04/26/2023] [Indexed: 05/22/2023]
Abstract
Agonist-induced GPCR phosphorylation is a key determinant for the binding and activation of β-arrestins (βarrs). However, it is not entirely clear how different GPCRs harboring divergent phosphorylation patterns impart converging active conformation on βarrs leading to broadly conserved functional responses such as desensitization, endocytosis, and signaling. Here, we present multiple cryo-EM structures of activated βarrs in complex with distinct phosphorylation patterns derived from the carboxyl terminus of different GPCRs. These structures help identify a P-X-P-P type phosphorylation motif in GPCRs that interacts with a spatially organized K-K-R-R-K-K sequence in the N-domain of βarrs. Sequence analysis of the human GPCRome reveals the presence of this phosphorylation pattern in a large number of receptors, and its contribution in βarr activation is demonstrated by targeted mutagenesis experiments combined with an intrabody-based conformational sensor. Taken together, our findings provide important structural insights into the ability of distinct GPCRs to activate βarrs through a significantly conserved mechanism.
Collapse
Affiliation(s)
- Jagannath Maharana
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Parishmita Sarma
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Manish K Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Sayantan Saha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Vinay Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Shirsha Saha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Mohamed Chami
- BioEM Lab, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Ramanuj Banerjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India.
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India.
| |
Collapse
|
33
|
Grimes J, Koszegi Z, Lanoiselée Y, Miljus T, O'Brien SL, Stepniewski TM, Medel-Lacruz B, Baidya M, Makarova M, Mistry R, Goulding J, Drube J, Hoffmann C, Owen DM, Shukla AK, Selent J, Hill SJ, Calebiro D. Plasma membrane preassociation drives β-arrestin coupling to receptors and activation. Cell 2023; 186:2238-2255.e20. [PMID: 37146613 PMCID: PMC7614532 DOI: 10.1016/j.cell.2023.04.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 12/16/2022] [Accepted: 04/12/2023] [Indexed: 05/07/2023]
Abstract
β-arrestin plays a key role in G protein-coupled receptor (GPCR) signaling and desensitization. Despite recent structural advances, the mechanisms that govern receptor-β-arrestin interactions at the plasma membrane of living cells remain elusive. Here, we combine single-molecule microscopy with molecular dynamics simulations to dissect the complex sequence of events involved in β-arrestin interactions with both receptors and the lipid bilayer. Unexpectedly, our results reveal that β-arrestin spontaneously inserts into the lipid bilayer and transiently interacts with receptors via lateral diffusion on the plasma membrane. Moreover, they indicate that, following receptor interaction, the plasma membrane stabilizes β-arrestin in a longer-lived, membrane-bound state, allowing it to diffuse to clathrin-coated pits separately from the activating receptor. These results expand our current understanding of β-arrestin function at the plasma membrane, revealing a critical role for β-arrestin preassociation with the lipid bilayer in facilitating its interactions with receptors and subsequent activation.
Collapse
Affiliation(s)
- Jak Grimes
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK
| | - Zsombor Koszegi
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK
| | - Yann Lanoiselée
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK
| | - Tamara Miljus
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK
| | - Shannon L O'Brien
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK
| | - Tomasz M Stepniewski
- Research Program on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, 08003, Spain
| | - Brian Medel-Lacruz
- Research Program on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, 08003, Spain
| | - Mithu Baidya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Maria Makarova
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK; School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ravi Mistry
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK
| | - Joëlle Goulding
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Julia Drube
- Institut für Molekulare Zellbiologie, Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität, Jena 07745, Germany
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität, Jena 07745, Germany
| | - Dylan M Owen
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK; Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Jana Selent
- Research Program on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, 08003, Spain
| | - Stephen J Hill
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Davide Calebiro
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
34
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
35
|
Kaur S, Sokrat B, Capozzi ME, El K, Bai Y, Jazic A, Han B, Krishnakumar K, D'Alessio DA, Campbell JE, Bouvier M, Shenoy SK. The Ubiquitination Status of the Glucagon Receptor determines Signal Bias. J Biol Chem 2023; 299:104690. [PMID: 37037304 DOI: 10.1016/j.jbc.2023.104690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
The pancreatic hormone glucagon activates the glucagon receptor (GCGR), a class B seven-transmembrane G protein-coupled receptor (GPCR) that couples to the stimulatory heterotrimeric Gs protein and provokes protein kinase A-dependent signaling cascades vital to hepatic glucose metabolism and islet insulin secretion. Glucagon-stimulation also initiates recruitment of the endocytic adaptors, β-arrestin1 and β-arrestin2, which regulate desensitization and internalization of the GCGR. Unlike many other GPCRs, the GCGR expressed at the plasma membrane is constitutively ubiquitinated and upon agonist-activation, internalized GCGRs are deubiquitinated at early endosomes and recycled via Rab4-containing vesicles. Herein we report a novel link between the ubiquitination status and signal transduction mechanism of the GCGR. In the deubiquitinated state, coupling of the GCGR to Gs is diminished, while binding to β-arrestin is enhanced with signaling biased to a β-arrestin1-dependent p38 mitogen activated protein kinase (MAPK) pathway. This ubiquitin-dependent signaling bias arises through the modification of lysine333 (K333) on the cytoplasmic face of transmembrane helix V. Compared with the GCGR-WT, the mutant GCGR-K333R has impaired ubiquitination, diminished G protein coupling and protein kinase A signaling, but unimpaired potentiation of glucose-stimulated-insulin secretion in response to agonist-stimulation, which involves p38 MAPK signaling. Both WT and GCGR-K333R promote the formation of glucagon-induced β-arrestin1-dependent p38 signaling scaffold that requires canonical upstream MAPK-Kinase3, but is independent of Gs, Gi and β-arrestin2. Thus ubiquitination/deubiquitination at K333 in the GCGR defines the activation of distinct transducers with the potential to influence various facets of glucagon signaling in health and disease.
Collapse
Affiliation(s)
- Suneet Kaur
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Badr Sokrat
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, H3T 1J4 Canada; Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, H3T 1J4 Canada
| | - Megan E Capozzi
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Kimberley El
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Yushi Bai
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Aeva Jazic
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Bridgette Han
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Kaavya Krishnakumar
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford CA 94305
| | - David A D'Alessio
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Jonathan E Campbell
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, H3T 1J4 Canada; Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, H3T 1J4 Canada
| | - Sudha K Shenoy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
36
|
Kim KM. Unveiling the Differences in Signaling and Regulatory Mechanisms between Dopamine D2 and D3 Receptors and Their Impact on Behavioral Sensitization. Int J Mol Sci 2023; 24:ijms24076742. [PMID: 37047716 PMCID: PMC10095578 DOI: 10.3390/ijms24076742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023] Open
Abstract
Dopamine receptors are classified into five subtypes, with D2R and D3R playing a crucial role in regulating mood, motivation, reward, and movement. Whereas D2R are distributed widely across the brain, including regions responsible for motor functions, D3R are primarily found in specific areas related to cognitive and emotional functions, such as the nucleus accumbens, limbic system, and prefrontal cortex. Despite their high sequence homology and similar signaling pathways, D2R and D3R have distinct regulatory properties involving desensitization, endocytosis, posttranslational modification, and interactions with other cellular components. In vivo, D3R is closely associated with behavioral sensitization, which leads to increased dopaminergic responses. Behavioral sensitization is believed to result from D3R desensitization, which removes the inhibitory effect of D3R on related behaviors. Whereas D2R maintains continuous signal transduction through agonist-induced receptor phosphorylation, arrestin recruitment, and endocytosis, which recycle and resensitize desensitized receptors, D3R rarely undergoes agonist-induced endocytosis and instead is desensitized after repeated agonist exposure. In addition, D3R undergoes more extensive posttranslational modifications, such as glycosylation and palmitoylation, which are needed for its desensitization. Overall, a series of biochemical settings more closely related to D3R could be linked to D3R-mediated behavioral sensitization.
Collapse
Affiliation(s)
- Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea
| |
Collapse
|
37
|
Zhang L, Zhou Y, Wang Y, Yang L, Wang Y, Shan Z, Liang J, Xiao Z. Inhibiting PAC1 receptor internalization and endosomal ERK pathway activation may ameliorate hyperalgesia in a chronic migraine rat model. Cephalalgia 2023; 43:3331024231163131. [PMID: 36946245 DOI: 10.1177/03331024231163131] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
BACKGROUND Pituitary adenylate cyclase-activating polypeptide (PACAP) is a multipotent neuropeptide widely distributed in the trigeminovascular system (TVS) and higher brain regions. At present, the underlying mechanism of PACAP/PACAP type1 (PAC1) receptor in migraine generation remains unclear. METHODS The rat model of chronic migraine (CM) was established by repeated intraperitoneal injection of nitroglycerin (NTG). Von Frey filaments and hot plate tests were used to measure the mechanical and thermal thresholds. The expression levels of c-Fos, calcitonin gene-related peptide (CGRP), PACAP, PAC1, protein kinase A (PKA) and phosphorylated extracellular signal-regulated kinase (ERK) were assessed by western blotting or immunofluorescence staining. The internalization of PAC1 receptor was visualized by fluorescence microscope and laser scanning confocal microscope. RESULTS The results showed that c-Fos and CGRP expression significantly increased after repeated administrations of NTG or PACAP. Pitstop2 notably improved hyperalgesia in CM rats, while PACAP6-38 offered no benefit. In addition, PACAP-induced PAC1 receptor internalization, PKA and ERK pathways activation were blocked by Pitstop2 instead of PACAP6-38. CONCLUSIONS Our results demonstrate that inhibition of PAC1 receptor internalization could effectively improve allodynia in CM rats by restraining ERK signaling pathway activation in a chronic migraine rat model. Modulation of receptor internalization may be a novel perspective to explore specific mechanisms of PACAP signaling activation in the trigeminal vascular system.
Collapse
Affiliation(s)
- Lily Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanjie Zhou
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yajuan Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liu Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yue Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhengming Shan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Liang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheman Xiao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
38
|
Lewis V, Bonniwell EM, Lanham JK, Ghaffari A, Sheshbaradaran H, Cao AB, Calkins MM, Bautista-Carro MA, Arsenault E, Telfer A, Taghavi-Abkuh FF, Malcolm NJ, El Sayegh F, Abizaid A, Schmid Y, Morton K, Halberstadt AL, Aguilar-Valles A, McCorvy JD. A non-hallucinogenic LSD analog with therapeutic potential for mood disorders. Cell Rep 2023; 42:112203. [PMID: 36884348 PMCID: PMC10112881 DOI: 10.1016/j.celrep.2023.112203] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/30/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Hallucinations limit widespread therapeutic use of psychedelics as rapidly acting antidepressants. Here we profiled the non-hallucinogenic lysergic acid diethylamide (LSD) analog 2-bromo-LSD (2-Br-LSD) at more than 33 aminergic G protein-coupled receptors (GPCRs). 2-Br-LSD shows partial agonism at several aminergic GPCRs, including 5-HT2A, and does not induce the head-twitch response (HTR) in mice, supporting its classification as a non-hallucinogenic 5-HT2A partial agonist. Unlike LSD, 2-Br-LSD lacks 5-HT2B agonism, an effect linked to cardiac valvulopathy. Additionally, 2-Br-LSD produces weak 5-HT2A β-arrestin recruitment and internalization in vitro and does not induce tolerance in vivo after repeated administration. 2-Br-LSD induces dendritogenesis and spinogenesis in cultured rat cortical neurons and increases active coping behavior in mice, an effect blocked by the 5-HT2A-selective antagonist volinanserin (M100907). 2-Br-LSD also reverses the behavioral effects of chronic stress. Overall, 2-Br-LSD has an improved pharmacological profile compared with LSD and may have profound therapeutic value for mood disorders and other indications.
Collapse
Affiliation(s)
- Vern Lewis
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Emma M Bonniwell
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Janelle K Lanham
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Abdi Ghaffari
- BetterLife Pharma Inc., Vancouver, BC V6H 1A6, Canada
| | | | - Andrew B Cao
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Maggie M Calkins
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Emily Arsenault
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Andre Telfer
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| | | | - Nicholas J Malcolm
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Fatema El Sayegh
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Yasmin Schmid
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kathleen Morton
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam L Halberstadt
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | - John D McCorvy
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
39
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
40
|
Kumar R, Milanesi S, Szpakowska M, Dotta L, Di Silvestre D, Trotta AM, Bello AM, Giacomelli M, Benedito M, Azevedo J, Pereira A, Cortesao E, Vacchini A, Castagna A, Pinelli M, Moratto D, Bonecchi R, Locati M, Scala S, Chevigné A, Borroni EM, Badolato R. Reduced G protein signaling despite impaired internalization and β-arrestin recruitment in patients carrying a CXCR4Leu317fsX3 mutation causing WHIM syndrome. JCI Insight 2023; 8:145688. [PMID: 36883568 PMCID: PMC10077478 DOI: 10.1172/jci.insight.145688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/25/2023] [Indexed: 03/09/2023] Open
Abstract
WHIM syndrome is an inherited immune disorder caused by an autosomal dominant heterozygous mutation in CXCR4. The disease is characterized by neutropenia/leukopenia (secondary to retention of mature neutrophils in bone marrow), recurrent bacterial infections, treatment-refractory warts, and hypogammaglobulinemia. All mutations reported in WHIM patients lead to the truncations in the C-terminal domain of CXCR4, R334X being the most frequent. This defect prevents receptor internalization and enhances both calcium mobilization and ERK phosphorylation, resulting in increased chemotaxis in response to the unique ligand CXCL12. Here, we describe 3 patients presenting neutropenia and myelokathexis, but normal lymphocyte count and immunoglobulin levels, carrying what we believe to be a novel Leu317fsX3 mutation in CXCR4, leading to a complete truncation of its intracellular tail. The analysis of the L317fsX3 mutation in cells derived from patients and in vitro cellular models reveals unique signaling features in comparison with R334X mutation. The L317fsX3 mutation impairs CXCR4 downregulation and β-arrestin recruitment in response to CXCL12 and reduces other signaling events - including ERK1/2 phosphorylation, calcium mobilization, and chemotaxis - all processes that are typically enhanced in cells carrying the R334X mutation. Our findings suggest that, overall, the L317fsX3 mutation may be causative of a form of WHIM syndrome not associated with an augmented CXCR4 response to CXCL12.
Collapse
Affiliation(s)
- Rajesh Kumar
- "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Brescia, Italy.,Rheumatology and Clinical Immunology, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy
| | - Samantha Milanesi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Laura Dotta
- "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Brescia, Italy.,Department of Pediatrics, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Dario Di Silvestre
- Institute for Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Anna Maria Trotta
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Anna Maria Bello
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Mauro Giacomelli
- "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Brescia, Italy
| | - Manuela Benedito
- Department of Clinical Hematology, Pediatric Hospital, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Joana Azevedo
- Department of Clinical Hematology, Pediatric Hospital, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Alexandra Pereira
- Department of Clinical Hematology, Pediatric Hospital, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Emilia Cortesao
- Department of Clinical Hematology, Pediatric Hospital, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | | | - Marinella Pinelli
- "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Brescia, Italy
| | - Daniele Moratto
- "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Bonecchi
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Massimo Locati
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Elena M Borroni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Raffaele Badolato
- "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Brescia, Italy.,Department of Pediatrics, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
41
|
An Update of G-Protein-Coupled Receptor Signaling and Its Deregulation in Gastric Carcinogenesis. Cancers (Basel) 2023; 15:cancers15030736. [PMID: 36765694 PMCID: PMC9913146 DOI: 10.3390/cancers15030736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) belong to a cell surface receptor superfamily responding to a wide range of external signals. The binding of extracellular ligands to GPCRs activates a heterotrimeric G protein and triggers the production of numerous secondary messengers, which transduce the extracellular signals into cellular responses. GPCR signaling is crucial and imperative for maintaining normal tissue homeostasis. High-throughput sequencing analyses revealed the occurrence of the genetic aberrations of GPCRs and G proteins in multiple malignancies. The altered GPCRs/G proteins serve as valuable biomarkers for early diagnosis, prognostic prediction, and pharmacological targets. Furthermore, the dysregulation of GPCR signaling contributes to tumor initiation and development. In this review, we have summarized the research progress of GPCRs and highlighted their mechanisms in gastric cancer (GC). The aberrant activation of GPCRs promotes GC cell proliferation and metastasis, remodels the tumor microenvironment, and boosts immune escape. Through deep investigation, novel therapeutic strategies for targeting GPCR activation have been developed, and the final aim is to eliminate GPCR-driven gastric carcinogenesis.
Collapse
|
42
|
Yadav MK, Singh V, Saha S, Shukla AK. A streamlined protocol for expression and purification of wild-type β-arrestins. Methods Enzymol 2023; 682:465-475. [PMID: 36948711 DOI: 10.1016/bs.mie.2022.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The two isoforms of β-arrestins namely β-arrestin 1 and 2 interact with, and regulate a broad repertoire of G protein-coupled receptors (GPCRs). While several protocols have been described in the literature for purification of β-arrestins for biochemical and biophysical studies, some of these protocols involve multiple complicated steps that prolong the process and yield relatively smaller amounts of purified proteins. Here, we describe a simplified and streamlined protocol for expression and purification of β-arrestins using E. coli as an expression host. This protocol is based on N-terminal fusion of GST tag and involves a two-step protocol involving GST-based affinity chromatography and size exclusion chromatography. The protocol described here yields sufficient amounts of high-quality purified β-arrestins suitable for biochemical and structural studies.
Collapse
Affiliation(s)
- Manish K Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Vinay Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Sayantan Saha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India.
| |
Collapse
|
43
|
Liu J, Yin Y, Ni J, Zhang P, Li WM, Liu Z. Dual Specific Phosphatase 7 Exacerbates Dilated Cardiomyopathy, Heart Failure, and Cardiac Death by Inactivating the ERK1/2 Signaling Pathway. J Cardiovasc Transl Res 2022; 15:1219-1238. [PMID: 35596107 DOI: 10.1007/s12265-022-10268-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/25/2022] [Indexed: 12/16/2022]
Abstract
Heart failure is one of the most common but complicated end-stage syndromes in clinical practice. Dilated cardiomyopathy is a myocardial structural abnormality that is associated with heart failure. Dual-specificity phosphatases (DUSPs) are a group of protein phosphatases that regulate signaling pathways in numerous diseases; however, their physiological and pathological impact on cardiovascular disease remains unknown. In the present study, we generated two transgenic mouse models, a DUSP7 knockout and a cardiac-specific DUSP7 overexpressor. Mice overexpressing DUSP7 showed an exacerbated disease phenotype, including severe dilated cardiomyopathy, heart failure, and cardiac death. We further demonstrated that high levels of DUSP7 inhibited ERK1/2 phosphorylation and influenced downstream c-MYC, c-FOS, and c-JUN gene expression but did not affect upstream activators. Taken together, our study reveals a novel molecular mechanism for DUSP7 and provides a new therapeutic target and clinical path to alleviate dilated cardiomyopathy and improve cardiac function.
Collapse
Affiliation(s)
- Jing Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yihen Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Heart, Lung, and Blood Center, Pan-Vascular Research Institute, Tongji University School of Medicine, Shanghai, China
| | - Jing Ni
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peiyu Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei-Ming Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
- Heart, Lung, and Blood Center, Pan-Vascular Research Institute, Tongji University School of Medicine, Shanghai, China.
| | - Zheng Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
- Heart, Lung, and Blood Center, Pan-Vascular Research Institute, Tongji University School of Medicine, Shanghai, China.
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Guangdong Province, Shenzhen, China.
| |
Collapse
|
44
|
Lala T, Doan JK, Takatsu H, Hartzell HC, Shin HW, Hall RA. Phosphatidylserine exposure modulates adhesion GPCR BAI1 (ADGRB1) signaling activity. J Biol Chem 2022; 298:102685. [PMID: 36370845 PMCID: PMC9723945 DOI: 10.1016/j.jbc.2022.102685] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/10/2022] Open
Abstract
Brain-specific angiogenesis inhibitor 1 (BAI1; also called ADGRB1 or B1) is an adhesion G protein-coupled receptor known from studies on macrophages to bind to phosphatidylserine (PS) on apoptotic cells via its N-terminal thrombospondin repeats. A separate body of work has shown that B1 regulates postsynaptic function and dendritic spine morphology via signaling pathways involving Rac and Rho. However, it is unknown if PS binding by B1 has any effect on the receptor's signaling activity. To shed light on this subject, we studied G protein-dependent signaling by B1 in the absence and presence of coexpression with the PS flippase ATP11A in human embryonic kidney 293T cells. ATP11A expression reduced the amount of PS exposed extracellularly and also strikingly reduced the signaling activity of coexpressed full-length B1 but not a truncated version of the receptor lacking the thrombospondin repeats. Further experiments with an inactive mutant of ATP11A showed that the PS flippase function of ATP11A was required for modulation of B1 signaling. In coimmunoprecipitation experiments, we made the surprising finding that ATP11A not only modulates B1 signaling but also forms complexes with B1. Parallel studies in which PS in the outer leaflet was reduced by an independent method, deletion of the gene encoding the endogenous lipid scramblase anoctamin 6 (ANO6), revealed that this manipulation also markedly reduced B1 signaling. These findings demonstrate that B1 signaling is modulated by PS exposure and suggest a model in which B1 serves as a PS sensor at synapses and in other cellular contexts.
Collapse
Affiliation(s)
- Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Juleva K Doan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hiroyuki Takatsu
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hye-Won Shin
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
45
|
Janetzko J, Kise R, Barsi-Rhyne B, Siepe DH, Heydenreich FM, Kawakami K, Masureel M, Maeda S, Garcia KC, von Zastrow M, Inoue A, Kobilka BK. Membrane phosphoinositides regulate GPCR-β-arrestin complex assembly and dynamics. Cell 2022; 185:4560-4573.e19. [PMID: 36368322 PMCID: PMC10030194 DOI: 10.1016/j.cell.2022.10.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/22/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022]
Abstract
Binding of arrestin to phosphorylated G protein-coupled receptors (GPCRs) is crucial for modulating signaling. Once internalized, some GPCRs remain complexed with β-arrestins, while others interact only transiently; this difference affects GPCR signaling and recycling. Cell-based and in vitro biophysical assays reveal the role of membrane phosphoinositides (PIPs) in β-arrestin recruitment and GPCR-β-arrestin complex dynamics. We find that GPCRs broadly stratify into two groups, one that requires PIP binding for β-arrestin recruitment and one that does not. Plasma membrane PIPs potentiate an active conformation of β-arrestin and stabilize GPCR-β-arrestin complexes by promoting a fully engaged state of the complex. As allosteric modulators of GPCR-β-arrestin complex dynamics, membrane PIPs allow for additional conformational diversity beyond that imposed by GPCR phosphorylation alone. For GPCRs that require membrane PIP binding for β-arrestin recruitment, this provides a mechanism for β-arrestin release upon translocation of the GPCR to endosomes, allowing for its rapid recycling.
Collapse
Affiliation(s)
- John Janetzko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Benjamin Barsi-Rhyne
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA; Department of Psychiatry, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA
| | - Dirk H Siepe
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Franziska M Heydenreich
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Matthieu Masureel
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shoji Maeda
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark von Zastrow
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA; Department of Psychiatry, University of California, San Francisco, School of Medicine, San Francisco, CA 94158, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan.
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
46
|
Jong YI, Harmon SK, O'Malley KL. GPCR
Signaling from Intracellular Membranes. GPCRS AS THERAPEUTIC TARGETS 2022:216-298. [DOI: 10.1002/9781119564782.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
47
|
Noradrenergic consolidation of social recognition memory is mediated by β-arrestin-biased signaling in the mouse prefrontal cortex. Commun Biol 2022; 5:1097. [PMID: 36253525 PMCID: PMC9576713 DOI: 10.1038/s42003-022-04051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2022] Open
Abstract
Social recognition memory (SRM) is critical for maintaining social relationships and increasing the survival rate. The medial prefrontal cortex (mPFC) is an important brain area associated with SRM storage. Norepinephrine (NE) release regulates mPFC neuronal intrinsic excitability and excitatory synaptic transmission, however, the roles of NE signaling in the circuitry of the locus coeruleus (LC) pathway to the mPFC during SRM storage are unknown. Here we found that LC-mPFC NE projections bidirectionally regulated SRM consolidation. Propranolol infusion and β-adrenergic receptors (β-ARs) or β-arrestin2 knockout in the mPFC disrupted SRM consolidation. When carvedilol, a β-blocker that can mildly activate β-arrestin-biased signaling, was injected, the mice showed no significant suppression of SRM consolidation. The impaired SRM consolidation caused by β1-AR or β-arrestin2 knockout in the mPFC was not rescued by activating LC-mPFC NE projections; however, the impaired SRM by inhibition of LC-mPFC NE projections or β1-AR knockout in the mPFC was restored by activating the β-arrestin signaling pathway in the mPFC. Furthermore, the activation of β-arrestin signaling improved SRM consolidation in aged mice. Our study suggests that LC-mPFC NE projections regulate SRM consolidation through β-arrestin-biased β-AR signaling. Social memory consolidation requires norepinephrine release in the medial prefrontal cortex (mPFC), and enhancing beta-arrestin signaling in the mPFC restores social recognition memory that is normally impaired by age in mice.
Collapse
|
48
|
Lala T, Hall RA. Adhesion G protein-coupled receptors: structure, signaling, physiology, and pathophysiology. Physiol Rev 2022; 102:1587-1624. [PMID: 35468004 PMCID: PMC9255715 DOI: 10.1152/physrev.00027.2021] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/11/2022] [Accepted: 04/16/2022] [Indexed: 01/17/2023] Open
Abstract
Adhesion G protein-coupled receptors (AGPCRs) are a family of 33 receptors in humans exhibiting a conserved general structure but diverse expression patterns and physiological functions. The large NH2 termini characteristic of AGPCRs confer unique properties to each receptor and possess a variety of distinct domains that can bind to a diverse array of extracellular proteins and components of the extracellular matrix. The traditional view of AGPCRs, as implied by their name, is that their core function is the mediation of adhesion. In recent years, though, many surprising advances have been made regarding AGPCR signaling mechanisms, activation by mechanosensory forces, and stimulation by small-molecule ligands such as steroid hormones and bioactive lipids. Thus, a new view of AGPCRs has begun to emerge in which these receptors are seen as massive signaling platforms that are crucial for the integration of adhesive, mechanosensory, and chemical stimuli. This review article describes the recent advances that have led to this new understanding of AGPCR function and also discusses new insights into the physiological actions of these receptors as well as their roles in human disease.
Collapse
Affiliation(s)
- Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
49
|
Komolafe K, Pacurari M. CXC Chemokines in the Pathogenesis of Pulmonary Disease and Pharmacological Relevance. Int J Inflam 2022; 2022:4558159. [PMID: 36164329 PMCID: PMC9509283 DOI: 10.1155/2022/4558159] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Chemokines and their receptors play important roles in the pathophysiology of many diseases by regulating the cellular migration of major inflammatory and immune players. The CXC motif chemokine subfamily is the second largest family, and it is further subdivided into ELR motif CXC (ELR+) and non-ELR motif (ELR-) CXC chemokines, which are effective chemoattractants for neutrophils and lymphocytes/monocytes, respectively. These chemokines and their receptors are expected to have a significant impact on a wide range of lung diseases, many of which have inflammatory or immunological underpinnings. As a result, manipulations of this subfamily of chemokines and their receptors using small molecular agents and other means have been explored for potential therapeutic benefit in the setting of several lung pathologies. Furthermore, encouraging preclinical data has necessitated the progression of a few of these drugs into clinical trials in order to make the most effective use of interventions in the development of viable targeted therapeutics. The current review presents the understanding of the roles of CXC ligands (CXCLs) and their cognate receptors (CXCRs) in the pathogenesis of several lung diseases such as allergic rhinitis, COPD, lung fibrosis, lung cancer, pneumonia, and tuberculosis. The potential therapeutic benefits of pharmacological or other CXCL/CXCR axis manipulations are also discussed.
Collapse
Affiliation(s)
- Kayode Komolafe
- RCMI Center for Health Disparities Research, Jackson State University, Jackson, MS 39217, USA
| | - Maricica Pacurari
- RCMI Center for Health Disparities Research, Jackson State University, Jackson, MS 39217, USA
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, Jackson, MS 39217, USA
| |
Collapse
|
50
|
Emerging Roles of the Nervous System in Gastrointestinal Cancer Development. Cancers (Basel) 2022; 14:cancers14153722. [PMID: 35954387 PMCID: PMC9367305 DOI: 10.3390/cancers14153722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Nerve–cancer cross-talk has increasingly become a focus of the oncology field, particularly in gastrointestinal (GI) cancers. The indispensable roles of the nervous system in GI tumorigenesis and malignancy have been dissected by epidemiological, experimental animal and mechanistic data. Herein, we review and integrate recent discoveries linking the nervous system to GI cancer initiation and progression, and focus on the molecular mechanisms by which nerves and neural receptor pathways drive GI malignancy. Abstract Our understanding of the fascinating connection between nervous system and gastrointestinal (GI) tumorigenesis has expanded greatly in recent years. Recent studies revealed that neurogenesis plays an active part in GI tumor initiation and progression. Tumor-driven neurogenesis, as well as neurite outgrowth of the pre-existing peripheral nervous system (PNS), may fuel GI tumor progression via facilitating cancer cell proliferation, chemoresistance, invasion and immune escape. Neurotransmitters and neuropeptides drive the activation of various oncogenic pathways downstream of neural receptors within cancer cells, underscoring the importance of neural signaling pathways in GI tumor malignancy. In addition, neural infiltration also plays an integral role in tumor microenvironments, and contributes to an environment in favor of tumor angiogenesis, immune evasion and invasion. Blockade of tumor innervation via denervation or pharmacological agents may serve as a promising therapeutic strategy against GI tumors. In this review, we summarize recent findings linking the nervous system to GI tumor progression, set the spotlight on the molecular mechanisms by which neural signaling fuels cancer aggressiveness, and highlight the importance of targeting neural mechanisms in GI tumor therapy.
Collapse
|